Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1085-L1092, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33822656

ABSTRACT

Resolution of the acute respiratory distress syndrome (ARDS) from pneumonia requires repair of the injured lung endothelium and alveolar epithelium, removal of neutrophils from the distal airspaces of the lung, and clearance of the pathogen. Previous studies have demonstrated the importance of specialized proresolving mediators (SPMs) in the regulation of host responses during inflammation. Although ARDS is commonly caused by Streptococcus pneumoniae, the role of lipoxin A4 (LXA4) and resolvin D1 (RvD1) in pneumococcal pneumonia is not well understood. In the present experimental study, we tested the hypothesis that endogenous SPMs play a role in the resolution of lung injury in a clinically relevant model of bacterial pneumonia. Blockade of formyl peptide receptor 2 (ALX/FPR2), the receptor for LXA4 and RvD1, with the peptide WRW4 resulted in more pulmonary edema, greater protein accumulation in the air spaces, and increased bacteria accumulation in the air spaces and the blood. Inhibition of this receptor was also associated with decreased levels of proinflammatory cytokines. Even in the presence of antibiotic treatment, WRW4 inhibited the resolution of lung injury. In summary, these experiments demonstrated two novel findings: LXA4 and RvD1 contribute to the resolution of lung injury due to pneumococcal pneumonia, and the mechanism of their benefit likely includes augmenting bacterial clearance and reducing pulmonary edema via the restoration of lung alveolar-capillary barrier permeability.


Subject(s)
Acute Lung Injury/drug therapy , Docosahexaenoic Acids/antagonists & inhibitors , Lipoxins/antagonists & inhibitors , Pneumonia, Pneumococcal/drug therapy , Receptors, Lipoxin/drug effects , Acute Lung Injury/complications , Acute Lung Injury/immunology , Animals , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Permeability/drug effects , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/immunology , Receptors, Lipoxin/metabolism , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology
2.
Arthritis Rheumatol ; 69(5): 996-1008, 2017 05.
Article in English | MEDLINE | ID: mdl-27860453

ABSTRACT

OBJECTIVE: Pain is a major symptom of osteoarthritis (OA); currently available analgesics either do not provide adequate pain relief or are associated with serious side effects. The aim of this study was to investigate the therapeutic potential of targeting the resolvin receptor system to modify OA pain and pathology. METHODS: Gene expression of 2 resolvin receptors (ALX and ChemR23) was quantified in synovium and medial tibial plateau specimens obtained from patients with OA at the time of joint replacement surgery. Two models of OA joint pain were used for the mechanistic studies. Gene expression in the joint and central nervous system was quantified. The effects of exogenous administration of the D series resolvin precursor 17(R)-hydroxy-docosahexaenoic acid (17[R]-HDoHE) on pain behavior, joint pathology, spinal microglia, and astroglyosis were quantified. Plasma levels of relevant lipids, resolvin D2, 17(R)-HDoHE, and arachidonic acid, were determined in rats, using liquid chromatography tandem mass spectrometry. RESULTS: There was a positive correlation between resolvin receptor and interleukin-6 (IL-6) expression in human OA synovial and medial tibial plateau tissue. In rats, synovial expression of ALX was positively correlated with expression of IL-1ß, tumor necrosis factor, and cyclooxygenase 2. Treatment with 17(R)-HDoHE reversed established pain behavior (but not joint pathology) in 2 models of OA pain. This was associated with a significant elevation in the plasma levels of resolvin D2 and a significant reduction in astrogliosis in the spinal cord in the monosodium iodoacetate-induced OA rat model. CONCLUSION: Our preclinical data demonstrate the robust analgesic effects of activation of the D series resolvin pathways in 2 different animal models of OA. Our data support a predominant central mechanism of action in clinically relevant models of OA pain.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Behavior, Animal/drug effects , Docosahexaenoic Acids/pharmacology , Osteoarthritis, Knee/genetics , Receptors, Chemokine/genetics , Animals , Arthralgia/chemically induced , Cartilage, Articular/pathology , Disease Models, Animal , Enzyme Inhibitors/toxicity , Gene Expression , Humans , Iodoacetic Acid/toxicity , Menisci, Tibial/surgery , Neuroglia/cytology , Neuroglia/metabolism , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptors, Chemokine/drug effects , Receptors, Lipoxin/drug effects , Receptors, Lipoxin/genetics , Spinal Cord/cytology , Spinal Cord/metabolism , Synovial Membrane/pathology
3.
Physiol Rep ; 4(19)2016 10.
Article in English | MEDLINE | ID: mdl-27694530

ABSTRACT

Sjögren's syndrome (SS) is an autoimmune disorder characterized by chronic inflammation and destruction of salivary and lacrimal glands leading to dry mouth and dry eyes, respectively. Currently, the etiology of SS is unknown and the current therapies have no permanent benefit; therefore, new approaches are necessary to effectively treat this condition. Resolvins are highly potent endogenous lipid mediators that are synthesized during the resolution of inflammation to restore tissue homeostasis. Previous studies indicate that the resolvin family member, RvD1, binds to the ALX/FPR2 receptor to block inflammatory signals caused by tumor necrosis factor-alpha (TNF-α) in the salivary epithelium. More recently, the corticosteroid, dexamethasone (DEX), was shown to be effective in reducing salivary gland inflammation. However, DEX, as with other corticosteroids, elicits adverse secondary effects that could be ameliorated when used in smaller doses. Therefore, we investigated whether the more stable aspirin-triggered (AT) epimer, AT-RvD1, combined with reduced doses of DEX is effective in treating TNF-α-mediated disruption of polarized rat parotid gland (Par-C10) epithelial cell clusters. Our results indicate that AT-RvD1 and DEX individually reduced TNF-α-mediated alteration in the salivary epithelium (i.e, maintained cell cluster formation, increased lumen size, reduced apoptosis, and preserved cell survival signaling responses) as compared to untreated cells. Furthermore, AT-RvD1 combined with a reduced dose of DEX produced stronger responses (i.e., robust salivary cell cluster formation, larger lumen sizes, further reduced apoptosis, and sustained survival signaling responses) as compared to those observed with individual treatments. These studies demonstrate that AT-RvD1 combined with DEX is highly effective in treating TNF-α-mediated disruption of salivary gland epithelium.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Combined Modality Therapy/methods , Dexamethasone/pharmacology , Docosahexaenoic Acids/pharmacology , Glucocorticoids/pharmacology , Salivary Glands/cytology , Sjogren's Syndrome/pathology , Tumor Necrosis Factor-alpha/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Dexamethasone/administration & dosage , Docosahexaenoic Acids/administration & dosage , Glucocorticoids/administration & dosage , Inflammation/metabolism , Rats , Receptors, Lipoxin/drug effects , Receptors, Lipoxin/metabolism , Salivary Glands/drug effects , Salivary Glands/pathology , Salivary Glands/ultrastructure , Signal Transduction/physiology , Sjogren's Syndrome/drug therapy , Sjogren's Syndrome/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Stroke ; 47(2): 490-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26732571

ABSTRACT

BACKGROUND AND PURPOSE: Lipoxin A4 (LXA4) has been reported to reduce inflammation in several neurological injury models. We studied the effects of LXA4 on neuroinflammation after subarachnoid hemorrhage (SAH) in a rat model. METHODS: Two hundred and thirty-eight Sprague-Dawley male rats, weight 280-320 g, were used. Exogenous LXA4 (0.3 and 1.0 nmol) were injected intracerebroventricularly at 1.5 hours after SAH. Neurological scores, brain water content, and blood-brain barrier were evaluated at 24 hours after SAH; Morris water maze and T-maze tests were examined at 21 days after SAH. The expression of endogenous LXA4 and its receptor formyl peptide receptor 2 (FPR2), as well as p38, interleukin-1ß, and interleukin-6 were studied either by ELISA or by Western blots. Neutrophil infiltration was observed by myeloperoxidase staining. FPR2 siRNA was used to knock down LXA4 receptor. RESULTS: The expression of endogenous LXA4 decreased, and the expression of FPR2 increased after SAH. Exogenous LXA4 decreased brain water content, reduced Evans blue extravasation, and improved neurological functions and improved the learning and memory ability after SAH. LXA4 reduced neutrophil infiltration and phosphorylation of p38, interleukin-1ß, and interleukin-6. These effects of LXA4 were abolished by FPR2 siRNA. CONCLUSIONS: Exogenous LXA4 inhibited inflammation by activating FPR2 and inhibiting p38 after SAH. LXA4 may serve as an alternative treatment to relieve early brain injury after SAH.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Lipoxins/pharmacology , Receptors, Lipoxin/drug effects , Subarachnoid Hemorrhage/immunology , p38 Mitogen-Activated Protein Kinases/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blotting, Western , Brain Edema/immunology , Brain Edema/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Inflammation , Injections, Intraventricular , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lipoxins/immunology , Lipoxins/metabolism , Male , Memory/drug effects , Neuropsychological Tests , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Rats , Rats, Sprague-Dawley , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Spatial Learning/drug effects , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Leukoc Biol ; 97(4): 689-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724390

ABSTRACT

Most staphylococci produce short α-type PSMs and about twice as long ß-type PSMs that are potent leukocyte attractants and toxins. PSMs are usually secreted with the N-terminal formyl group but are only weak agonists for the leukocyte FPR1. Instead, the FPR1-related FPR2 senses PSMs efficiently and is crucial for leukocyte recruitment in infection. Which structural features distinguish FPR1 from FPR2 ligands has remained elusive. To analyze which peptide properties may govern the capacities of ß-type PSMs to activate FPRs, full-length and truncated variants of such peptides from Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis were synthesized. FPR2 activation was observed even for short N- or C-terminal ß-type PSM variants once they were longer than 18 aa, and this activity increased with length. In contrast, the shortest tested peptides were potent FPR1 agonists, and this property declined with increasing peptide length. Whereas full-length ß-type PSMs formed α-helices and exhibited no FPR1-specific activity, the truncated peptides had less-stable secondary structures, were weak agonists for FPR1, and required N-terminal formyl-methionine residues to be FPR2 agonists. Together, these data suggest that FPR1 and FPR2 have opposed ligand preferences. Short, flexible PSM structures may favor FPR1 but not FPR2 activation, whereas longer peptides with α-helical, amphipathic properties are strong FPR2 but only weak FPR1 agonists. These findings should help to unravel the ligand specificities of 2 critical human PRRs, and they may be important for new, anti-infective and anti-inflammatory strategies.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Hemolysin Proteins/chemistry , Neutrophils/drug effects , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/drug effects , Amino Acid Sequence , Bacterial Proteins/chemical synthesis , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Calcium Signaling/drug effects , Chemotaxis, Leukocyte/drug effects , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , HL-60 Cells , Hemolysin Proteins/chemical synthesis , Hemolysin Proteins/pharmacology , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Molecular Sequence Data , N-Formylmethionine/chemistry , Neutrophils/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Folding , Protein Stability , Protein Structure, Secondary , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Staphylococcus aureus/chemistry , Staphylococcus epidermidis/chemistry , Staphylococcus lugdunensis/chemistry , Structure-Activity Relationship
6.
J Surg Res ; 183(2): 710-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23558258

ABSTRACT

BACKGROUND: Hemorrhagic shock activates cellular stress signals and can lead to systemic inflammatory response, organ injury, and death. Mitogen-activated protein kinase (MAPK) acts as a sensor of tissue injury in models of ischemia-reperfusion injury. Lipoxins are endogenous lipid mediators with potent anti-inflammatory and pro-resolving actions. We hypothesized that BML-111 (a lipoxin A4-receptor agonist) attenuates hemorrhagic shock-induced acute lung injury (ALI) through inhibiting activation of the MAPK pathway. METHODS: We randomized Sprague-Dawley rats into four groups: sham, hemorrhagic shock-resuscitation (HS), HS plus BML-111 (BML-111), and HS plus BML-111 and BOC-2 (BOC-2). Two hours after resuscitation, we collected samples of lung. We obtained bronchoalveolar lavage fluid for neutrophil count. We performed optical microscopy to examine pathologic changes in lungs. Wet/dry ratios, myeloperoxidase expression, interleukin (IL)-1ß and IL-6 levels in lung were measured. We evaluated MAPK activation and the DNA binding activity of activator protein-1 in lung. RESULTS: Treatment with BML-111 reduced the lung damage and wet/dry ratio, neutrophil count in bronchoalveolar lavage fluid, expression of myeloperoxidase, and production of IL-1ß and IL-6 in lung. Phosphorylation of MAPK was also decreased by BML-111 in lung. Furthermore, the DNA binding activity of activator protein-1 was blocked by BML-111. An antagonist of the lipoxin A4-receptor, BOC-2, reversed the protective effect of BML-111 on ALI induced by hemorrhagic shock. CONCLUSIONS: This study indicates that BML-111 attenuated hemorrhagic shock-induced ALI via the MAPK/activator protein-1 signaling pathway. Therefore, BML-111 may have therapeutic potential for hemorrhagic shock-induced ALI.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/prevention & control , Heptanoic Acids/therapeutic use , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Shock, Hemorrhagic/complications , Signal Transduction/physiology , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Heptanoic Acids/pharmacology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/physiology , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Lipoxin/agonists , Receptors, Lipoxin/antagonists & inhibitors , Receptors, Lipoxin/drug effects , Signal Transduction/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/physiology
7.
Arch Biochem Biophys ; 516(2): 121-7, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22005393

ABSTRACT

Calpain inhibitors, including peptide aldehydes (N-acetyl-Leu-Leu-Nle-CHO and N-acetyl-Leu-Leu-Met-CHO) and α-mercapto-acrylic acid derivatives (PD150606 and PD151746), have been shown to stimulate phagocyte functions via activation of human formyl peptide receptor (hFPR) and/or hFPR-like 1 (hFPRL1). Using the homology modeling of the receptors and the ligand docking simulation, here we show that these calpain inhibitors could bind to the putative N-formyl-Met-Leu-Phe (fMLF) binding site on hFPR and/or hFPRL1. The studies with HEK-293 cells stably expressing hFPR or hFPRL1 showed that the concentrations of calpain inhibitors required to induce an increase in cytoplasmic free Ca(2+) ([Ca(2+)](i)) was much higher (>100 folds) than those of fMLF and Trp-Lys-Tyr-Met-Val-D-Met (WKYMVm). HEK-293 cells expressing hFPR or hFPRL1 with the mutated fMLF binding site never exhibited the [Ca(2+)](i) response to calpain inhibitors. When the optimal concentrations of each stimulus were used, pretreatment of cells with fMLF or WKYMVm abolished an increase in [Ca(2+)](i) induced by calpain inhibitors as well as the same stimulus, whereas pretreatment of cells with calpain inhibitors significantly suppressed, but never abolished, the [Ca(2+)](i) response induced by fMLF or WKYMVm, suggesting that the binding affinity of the inhibitors to the putative fMLF binding site may be lower than that of fMLF or WKYMVm.


Subject(s)
Glycoproteins/pharmacology , Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/chemistry , Receptors, Lipoxin/drug effects , Acrylates/pharmacology , Base Sequence , Binding Sites , Calcium Signaling/drug effects , Computer Simulation , Cysteine Proteinase Inhibitors/pharmacology , DNA Primers/genetics , HEK293 Cells , Humans , Leupeptins/pharmacology , Ligands , Models, Molecular , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Oligopeptides/pharmacology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein
8.
ScientificWorldJournal ; 6: 1375-84, 2006 Oct 25.
Article in English | MEDLINE | ID: mdl-17072491

ABSTRACT

The anti-inflammatory actions of the nonapeptide antiflammin-2, identified by homology with uteroglobin and annexin-A1 sequences, have been described in some detail, yet its mechanisms of action remain elusive. Since recent data indicate an involvement of the formyl peptide receptor (FPR)-like 1 (or FPRL-1) in the effects of annexin-A1, we have tested here the effect of antiflammin-2 with respect to this receptor family. Using HEK-293 cells expressing either human FPR and FPRL-1, and an annexin-A1 peptide as tracer ([125I-Tyr]-Ac2-26), we found that antiflammin-2 competed for binding only at FPRL-1, and not FPR, with an approximate EC50 of 1 mM. In line with data produced for the full-length protein, genuine receptor activation by antiflammin-2 was confirmed by rapid phosphorylation of extracellular-regulated kinase 1 and 2. Finally, study of the neutrophil interaction with activated endothelium under flow demonstrated an inhibitory effect of antiflammin-2, thus providing functional support to a role for the antiflammin-2/FPRL-1 anti-inflammatory axis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/drug effects , Binding, Competitive , Cells, Cultured , Humans , Kidney/cytology , Kidney/embryology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology
9.
Inflamm Allergy Drug Targets ; 5(2): 107-14, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16613569

ABSTRACT

This review will emphasize the concept of anti-inflammation and propose that better understanding of the resolution phase of the inflammatory response organized by the host against inflammatory insults can lead to the identification of novel targets for drug development. Under the umbrella of anti-inflammation, we discuss here recent discoveries in the biology of annexin 1 and glucocorticoids. The fact that annexin 1 and another anti-inflammatory mediator, lipoxin A4, converge onto a specific membrane receptor (termed ALX) might help understanding the resolution phase of inflammation, and strengthen the use of this target for innovative drug development. In addition, glucocorticoids (GC), historically linked to annexin 1, are widely use in the clinical control of several pathologies with an inflammatory etiology, though their use is often burdened by several side effects. The development of new GC with more specific or improved mechanisms, e.g. the nitro-steroids, would go along this wave and, potentially, could be of large applicability. The two mediators/targets here illustrated are to be taken as examples of the clues that the study of anti-inflammation might give to the pharmaceutical industry for innovative drug discovery.


Subject(s)
Annexin A1/metabolism , Anti-Inflammatory Agents/metabolism , Glucocorticoids/metabolism , Nitric Oxide Donors/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lipoxins/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Prednisolone/analogs & derivatives , Prednisolone/metabolism , Prednisolone/pharmacology , Prednisolone/therapeutic use , Receptors, Formyl Peptide/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Lipoxin/drug effects
10.
Int J Immunopathol Pharmacol ; 17(2): 145-56, 2004.
Article in English | MEDLINE | ID: mdl-15171815

ABSTRACT

Lipoxin A4 (LXA4) is a potent eicosanoid that inhibits IL-1beta-induced activation of human fibroblast like synoviocytes (FLS) via the ALX4 receptor. Serum amyloid A (SAA) is an acute phase reactant with cytokine-like properties. SAA has been shown to bind the same seven transmembrane G protein-coupled receptor ligated by LXA4. Here we compared the inflammatory responses of lipid (LXA4) and peptide (SAA) ligands in human FLS via the shared ALX, and characterized their downstream signaling. LXA4 induced stimulation of tissue inhibitors of metalloproteinase-2, whereas SAA induced interleukin-8 and matrix metalloproteinase-3 production. SAA up-regulated NF-kappaB and AP-1 DNA binding activity, while LXA4 markedly inhibited these responses after IL-1beta stimulation. A human IL-8 promoter luciferase construct was transfected into CHO cells stably expressing ALXR in order to determine the role of NF-kappaB and/or AP-1 in the regulation of IL-8 gene expression. The NF-kappaB pathway proved to be the preeminent for the biological responses elicited by both ligands. These findings suggest that two endogenous molecules, targeting a common receptor, could participate in the pathogenesis of inflammatory arthritis by differentially regulating inflammatory responses in tissues expressing the ALXR.


Subject(s)
Amyloid/pharmacology , Interleukin-8/biosynthesis , Lipoxins/pharmacology , NF-kappa B/biosynthesis , Arthritis, Rheumatoid/pathology , Cells, Cultured , DNA/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Humans , Interleukin-8/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinases/metabolism , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/drug effects , Synovial Membrane/cytology , Synovial Membrane/drug effects , Tissue Inhibitor of Metalloproteinases/pharmacology , Transfection
11.
J Immunol ; 172(11): 7078-85, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15153530

ABSTRACT

Alzheimer's disease (AD) is characterized by overproduction of beta amyloid peptides in the brain with progressive loss of neuronal cells. The 42-aa form of the beta amyloid peptide (Abeta(42)) is implied as a major causative factor, because it is toxic to neurons and elicits inflammatory responses in the brain by activating microglial cells. Despite the overproduction of Abeta(42), AD brain tissue also generates protective factor(s) that may antagonize the neurodestructive effect of Abeta(42). Humanin is a gene cloned from an apparently normal region of an AD brain and encodes a 24-aa peptide. Both secreted and synthetic Humanin peptides protect neuronal cells from damage by Abeta(42), and the effect of Humanin may involve putative cellular receptor(s). To elucidate the molecular identity of such receptor(s), we examined the activity of synthetic Humanin on various cells and found that Humanin induced chemotaxis of mononuclear phagocytes by using a human G protein-coupled formylpeptide receptor-like-1 (FPRL1) and its murine counterpart FPR2. Coincidentally, FPRL1 and FPR2 are also functional receptors used by Abeta(42) to chemoattract and activate phagocytic cells. Humanin reduced the aggregation and fibrillary formation by suppressing the effect of Abeta(42) on mononuclear phagocytes. In neuroblast cells, Humanin and Abeta(42) both activated FPRL1; however, only Abeta(42) caused apoptotic death of the cells, and its cytopathic effect was blocked by Humanin. We conclude that Humanin shares human FPRL1 and mouse FPR2 with Abeta(42) and suggest that Humanin may exert its neuroprotective effects by competitively inhibiting the access of FPRL1 to Abeta(42).


Subject(s)
Neuroprotective Agents/pharmacology , Proteins/pharmacology , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/drug effects , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Humans , Intracellular Signaling Peptides and Proteins , Mice , Neurons/drug effects , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/agonists
12.
Br J Pharmacol ; 141(1): 37-46, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14662730

ABSTRACT

1. Human formyl peptide-receptor-like-1 (FPRL-1) is a promiscuous G protein-coupled receptor (GPCR), and belongs to a chemoattractant receptor family protein. This receptor has been reported to interact with various host-derived peptides and lipids involved in inflammatory responses. We described here, a novel role for FPRL-1 as a high-affinity beta-chemokine receptor for an N-terminally truncated form of the CKbeta8 (CCL23/MPIF-1) splice variant CKbeta8-1 (22-137 aa). 2. RT-PCR analysis of mRNA derived from human tissues and cells revealed a predominant expression of FPRL-1 in inflammatory cells, particularly in neutrophils. 3. Intracellular calcium mobilisation assay, used as screening tool, in recombinant Chinese hamster ovary (CHO-K1) and human embryonic kidney (HEK293s) cells coexpressing FPRL-1 and Galpha(16), demonstrated FPRL-1 is a functional high-affinity receptor for CKbeta8-1 (46-137 aa, sCKbeta8-1), with pEC(50) values of 9.13 and 8.85, respectively. 4. The FPRL-1 activation in CHO-K1 cells is mediated by Galpha(i)/Galpha(o) proteins, as assessed by pertussis toxin sensitivity and inhibition of forskolin-induced cyclic AMP accumulation. 5. Binding experiments were performed with a radio-iodinated synthetic peptide, [(125-)I]-WKYMVm, a known potent FPRL-1 agonist. CHO-K1 cell membranes expressing FPRL-1 bound [(125-)I]-WKYMVm with a K(d) value of 9.34. Many known FPRL-1 agonists were tested and sCKbeta8-1 was the most effective nonsynthetic ligand in displacing the radiolabelled agonist, with a pIC(50) of 7.97. 6. The functional significance of sCKbeta8-1 interaction with FPRL-1 was further demonstrated by the activation of polymorphonuclear leukocytes (PMNs) calcium mobilisation and chemotaxis. These interactions were shown to be via FPRL-1 by specific blockade of PMNs activation in the presence of an FPRL-1 antibody.


Subject(s)
Chemokines, CC/chemistry , Chemokines, CC/pharmacology , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/drug effects , Amino Acid Sequence , Animals , CHO Cells , Calcium/metabolism , Cell Movement/drug effects , Chemokines, CC/metabolism , Chemotaxis/drug effects , Cricetinae , Drug Evaluation, Preclinical/methods , Female , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression , Humans , Iodine Radioisotopes/metabolism , Kidney/cytology , Kidney/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...